HTLV-1 basic leucine zipper factor downregulates cyclin D1 expression via interactions with NF-κB

  • Authors:
    • Yunyun Ma
    • Bo Zhang
    • Dong Wang
    • Lili Qian
    • Xianmei Song
    • Xueyin Wang
    • Chaokuan Yang
    • Guoqiang Zhao
  • View Affiliations

  • Published online on: January 26, 2017     https://doi.org/10.3892/ijmm.2017.2868
  • Pages: 764-770
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Human T cell leukemia virus type 1 (HTLV-1) is an oncogenic retrovirus. It can cause adult T cell leukemia (ATL) and other diseases. The HTLV-1 basic leucine zipper (bZIP) factor (HBZ), which is encoded by the minus-strand of the provirus, is expressed in all cases of ATL and involved in T cell proliferation. However, the exact mechanism underlying its growth-promoting activity is poorly understood. Herein, we demonstrated that HBZ suppressed cyclin D1 expression by inhibiting the nuclear factor (NF)-κB signaling pathway. Among the potential mechanisms of cyclin D1 inhibition mediated by HBZ, we found that HBZ suppressed cyclin D1 promoter activity. Luciferase assay analysis revealed that HBZ repressed cyclin D1 promoter activity by suppressing NF-κB‑driven transcription mediated by the p65 subunit. Using an immunoprecipitation assay, we found that HBZ could bind to p65, but not p50. Finally, we showed that HBZ selectively interacted with p65 via its AD+bZIP domains. By suppressing cyclin D1 expression, HBZ can alter cell cycle progression of HTLV-1-infected cells, which may be critical for oncogenesis.

Introduction

Nuclear factor (NF)-κB is a transcription factor member of the Rel family. The Rel family includes five members, RelA/p65, RelB, c-Rel, p50 and p52 (13), which have sequence similarity to over ~300 amino acids in the amino-terminal half of these proteins. These Rel family members can form heterodimers or homodimers with a range of DNA-binding and activation potentials. Even though all family members contain a Rel homology domain that is required for DNA binding, dimerization, and nuclear localization, only RelA/p65 (hereafter referred to as p65), RelB and c-Rel, have a specific carboxy terminal transactivation domain that can regulate NF-κB-dependent gene expression (3,4). The most widely studied active form of NF-κB is a p65-p50 heterodimer, which is a main activator of gene transcription (5,6).

Although this transcription factor was first known as a regulator of inflammation and immune responses, recent studies show that NF-κB also regulates cell proliferation (7,8). The mechanism of proliferation regulation involves the stimulation of cyclin E/CDK2 activity and c-Myc expression (911), as well as other proliferation-associated genes, such as colony stimulating factor, platelet-derived growth factor, and cyclin D1 (4,1214).

Cyclin D1 is a member of the D-type cyclins. It regulates the G1/S phase transition of the cell cycle. Cyclin D1 is induced by mitogens and can dimerize with CDK4/CDK6 to phosphorylate retinoblastoma (Rb) protein, which then derepresses E2F transcriptional activity to allow entry into the S-phase (15,16). Analysis of the cyclin D1 promoter revealed the presence of two κB sites located at positions −840 and −33, which have been deemed to be κB1 and κB2 (12), respectively. Mutational analysis demonstrated that binding of the p65/p50 complex to the −33 κB2 site is important for cyclin D1 transcriptional regulation during the proliferative response induced by growth factors or serum or by stimulation via the Ras/Rac pathway (12,13,17).

Human T cell leukemia virus type 1 (HTLV-1) is a complex retrovirus that can cause adult T cell leukemia (ATL) (18,19). HTLV-1 encodes several unique proteins that can participate in viral infectivity, replication, persistence and transformation (20). Among these proteins, HTLV-1 basic leucine zipper (bZIP) factor (HBZ) expression is conserved in all ATL cells and is thought to be essential for leukemogenesis (21). Additionally, the HBZ gene can promote ATL cell proliferation (21). Recent studies have shown that HBZ can inhibit Tax-mediated NF-κB activation and suppress the transcription of various NF-κB target genes (22). These findings suggest that HBZ is involved in the NF-κB signaling pathway, and also contributes to the regulation of cyclin D1, an NF-κB target gene.

In a previous study, we showed that HBZ downregulated cyclin D1 expression via interactions with cAMP-response element binding protein CREB (23). To characterize other mechanisms of cyclin D1 suppression via HBZ, we utilized 293T and Jurkat cells in which HBZ was stably expressed and studied the effects of HBZ on cyclin D1 expression. We found that HBZ mediated the suppression of cyclin D1 by interacting with p65.

Materials and methods

Cell culture

Jurkat cells (purchased from the China Center for Type Culture Collection, Wuhan, China) were cultured in RPMI-1640 medium (HyClone, Logan, UT, USA). 293T cells (purchased from the China Center for Type Culture Collection) were cultured in Dulbecco's modified Eagle's medium (DMEM) (HyClone). Media were supplemented with 10% fetal bovine serum (FBS; HyClone), 2 mM glutamine, 50 mg/ml streptomycin, and 100 U/ml penicillin. Cells were synchronized by serum starvation for 18 h, followed by stimulation in medium containing 10% FBS for 12 h.

Lentiviral vector construction and transfection with the recombinant lentivirus

Full-length sHBZ (amino acids 1-209) and deletion mutants (sHBZ-AD, amino acids 1-73; sHBZ-basic leucine zipper domain (bZIP), amino acids 155-209; sHBZ-ΔAD, amino acids 74-209; sHBZ-ΔbZIP, amino acids 1-154; and sHBZ-AD+bZIP, amino acids (1-73)+(155-209) were generated by PCR from pET-29a-HBZ (provided by Dr H.J. Zhi; Uniformed Services University of the Health Sciences, Bethesda, MD, USA) with an HA epitope tagged at the C- or N-terminus (Fig. 1). These fragments were subcloned into a lentiviral vector (LV5) and were transfected into 293T cells using Lipofectamine™ 2000 (Invitrogen, Carlsbad, CA, USA) as reported previously (23). Infection or transduction with lentiviral vectors was also performed as described previously (23).

Synthesis of cDNA and reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from the stable lentiviral-infected 293T cells, Jurkat cells and uninfected control cells using Total RNA kit I (R6834-01; Omega, Norcross, GA, USA) according to the manufacturer's instructions. For cDNA synthesis, 2.5 µg total RNA was reverse transcribed using the RevertAid™ First-Strand cDNA Synthesis kit (K1621; Fermentas, Carlsbad, CA, USA). For RT-PCR, cDNA products were quantified with Power SYBR-Green PCR Master Mix and an ABI 7500 Fast Real-Time PCR system (Applied Biosystems, Carlsbad, CA, USA). Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) mRNA transcript levels were quantified to normalize the amount of cDNA that was loaded. The customized primers used were: cyclin D1 forward, 5′-GACCATCCCCCTGACGGCCGAG-3′ and reverse, 5′-CGCACGTCGGTGGGTGTGC-3′; and GAPDH forward, 5′-AGAGGCAGGGATGATGTTCTG-3′ and reverse, 5′-GACTCATGACCACAGTCCATGC-3′. All PCR experiments were repeated at least 3 times. A melting curve analysis was performed to ensure the specificity of the products after amplification. The median in each triplicate dataset was used to calculate the relative cyclin D1 mRNA concentration (ΔCt = Ct median mRNA − Ct median GAPDH), which was then converted to x-fold-changes (2−ΔΔCt) (24).

Construction of cyclin D1 promoter deletion mutants and luciferase assays

The deletion mutants of human cyclin D1 promoter that lacked AP1-, STAT-/SP1-, CREB- and NF-κB-binding sites (CD1 to CD5) were generated by PCR and then cloned into the vector pGL3-enhancer (Promega, Madison, WI, USA), as previously reported (23). Luciferase assays were performed using 293T and Jurkat cells, as described previously (23). All reporter assays were performed in triplicate and repeated in three independent experiments. Firefly luciferase values were normalized by Renilla luciferase values from pRL-TK (Promega).

Immunoprecipitation and immunoblotting

To examine the protein-protein interactions in the 293T cells, subconfluent cells were transfected with various combinations of expression vectors. Cells were lysed 48 h later in RIPA buffer containing 50 mM Tris-HCl, pH 8.0, 100 mM NaCl, 1 mM MgCl2, 1% Triton X-100, 0.5% Nonidet P-40 and protease inhibitors. Lysates were precleared by incubation with 20 µl suspension of 50% protein G-agarose (GE Healthcare Life Sciences, Uppsala, Sweden) for 1 h at 4°C. Precleared lysates were incubated with anti-HA (Y-11) or anti-NF-κB p65 (C-20) (both from Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) antibodies for 1 h at 4°C, and immune complexes were collected by incubation with protein G-agarose at 4°C for 1 h. After extensive washing, immunoprecipitated proteins were resolved by 12% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and analyzed by western blotting with anti-p65 (SC-109), anti-p50 (H-119), or anti-HA (F-7) (all from Santa Cruz Biotechnology, Inc.) antibodies. Membranes were developed with ECL (GE Healthcare Life Sciences). Other antibodies were as follows: anti-mouse IgG and anti-rabbit IgG (GE Healthcare Life Sciences).

Statistical analysis

All data are presented as the means ± standard deviation (SD) from at least 3 independent experiments. Statistical analyses were performed with SPSS (version 19.0; SPSS Inc., Chicago, IL, USA). Statistical significance was assessed by one-way analysis of variance (ANOVA) or the Student's t-test. Differences were considered statistically significant when P-values were <0.05.

Results

HBZ inhibits the expression of cyclin D1

To analyze the effect of HBZ on cyclin D1 expression, we first transduced 293T cells with a lentiviral vector carrying the HBZ gene and evaluated cyclin D1 levels by RT-qPCR and western blot analysis. The expression of HBZ resulted in reduced expression levels of cyclin D1 mRNA and protein (Fig. 2A and C). To further characterize this effect in T cells, we transduced HBZ into Jurkat cells using a lentiviral vector. As in 293T cells, HBZ expression resulted in reduced cyclin D1 mRNA and protein levels in the Jurkat cells as detected by RT-qPCR and western blot assays.

Figure 2

Repression of cyclin D1 gene expression by HBZ. Cells that stably expressed HBZ or its mutants were generated by lentiviral-mediated transfection of the HBZ genes (HBZ, HBZ-AD, HBZ-bZIP, HBZ-ΔAD, HBZ-ΔbZIP or HBZ-AD+bZP). 293T cells were transduced with either LV-HBZ, LV-HBZ-AD, LV-HBZ-bZIP, HBZ-ΔAD, HBZ-ΔbZIP or HBZ-AD+bZP at an MOI of 5. Jurkat cells were transduced with LV-HBZ at an MOI of 30. Cells were selected in liquid media containing 1 mg/ml puromycin. (A) Levels of cyclin D1 mRNA transcripts were reduced in the HBZ-expressing cells. Real-time PCR was performed to analyze the expression of cyclin D1 mRNA in untransduced (293T or Jurkat) cells and transduced (293T-HBZ or Jurkat-HBZ) cells; error bars represent standard deviation (SD). (B) Cyclin D1 mRNA transcript levels were reduced by the bZIP and bZIP+AD domains of HBZ. Stable expression of HBZ and its mutants in 293T cells and empty 293T cells was confirmed by real-time PCR, as in (A); error bars represent SD. (C) Cyclin D1 expression was reduced by HBZ. The blot shows that 50 µg total cell protein from each HBZ stably transfected cell line was resolved using 15% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and immunoblotted using cyclin D1, HA and β-tubulin antibodies. (D) Cyclin D1 expression was reduced by HBZ via its bZIP and bZIP+AD domains. Cell lysates were prepared from 293T cells expressing stable HBZ and its mutants and empty-vector transduced 293T cells; immunoblots were carried out as in (C). *P<0.01; NS, no significant difference.

HBZ has three domains, and each has one of the following distinct functions (Fig. 1): an activation domain (AD), a central domain (CD), and a bZIP domain (2527). To identify the portion of HBZ that is necessary to inhibit the expression of cyclin D1, we constructed deletion mutants (Fig. 1). 293T cells were transduced with the lentiviral vectors carrying each of the aforementioned HBZ mutants, followed by RT-qPCR and western blot assays. Two mutants (HBZ-bZIP and HBZ-AD+bZIP) repressed cyclin D1 mRNA and protein levels (Fig. 2B and D). By contrast, three other mutants (HBZ-AD, HBZ-ΔAD and HBZ-ΔbZIP) caused no significant changes in cyclin D1 mRNA and protein levels. These findings indicate that the bZIP domain, with or without an AD domain, is needed to suppress levels of cyclin D1 mRNA and protein.

HBZ suppresses cyclin D1 promoter activity through the CRE and NF-κB sites

Based on the ability of HBZ to downregulate cyclin D1 gene transcription, we assessed whether cyclin D1 promoter activity was under the control of HBZ. We constructed a cyclin D1 promoter reporter plasmid and co-transfected it into HBZ stably expressing Jurkat cells with a control plasmid, pRL-TK. Our findings revealed that HBZ reduced cyclin D1 promoter activity in Jurkat T cells (data not shown).

The cyclin D1 promoter has multiple transcription factor-binding sites, such as AP-1, E2F, NF-κB and Oct-1 (28). Extracellular signals act via signal transduction pathways to converge at binding sites to activate or inhibit promoter activity and regulate cell cycle progression. To identify cis-element(s) in the cyclin D1 promoter region that are responsive to HBZ, we transfected luciferase report constructs that contained cyclin D1 promoters of various lengths into empty 293T cells and stable HBZ-expressing 293T cells. The promoter activity of full-length pGL3-CD1/-1021 in 293T-HBZ cells was reduced by ~30% compared with the activity noted in the empty 293T cells (Fig. 3). These findings are in accord with the aforementioned studies carried out in the Jurkat human T cell line. When the AP1 site was deleted (pGL3-CD2/-874), the promoter activity was not reduced. Furthermore, deletion of the STAT and SP1 (pGL3-CD3/-99) sites also did not reduce promoter activity, and pGL3-CD3/-99, which contained CRE and NF-κB sites, had promoter activity that was similar to pGL3-CD1/-1021, which contained a full-length cyclin D1 promoter. However, when the CRE site was deleted (pGL3-CD4/-47), promoter activity was decreased to 50% of the pGL3-CD3/-99 activity in the empty 293T cells, while in the 293T-HBZ cells it decreased to 40%. The promoter activity of pGL3-CD4/-47 in the 293T-HBZ cells was reduced 20% compared with that noted in the empty 293T cells. Further deletion of the NF-κB site (pGL3-CD5/-26) resulted in reduction of the promoter activity to ~30% of pGL3-CD4/-47 in the empty 293T cells. By contrast, there was no significant difference in pGL3-CD5/-26 promoter activity between the 293T and 293T-HBZ cells. Although we previously reported that the CRE sites were critical for HBZ-mediated suppression of cyclin D1 promoter activity (23), these findings indicate that the NF-κB site is also responsible for HBZ-mediated suppression of cyclin D1 promoter activity.

Domains of HBZ that are responsible for NF-κB p65 inactivation

Previous studies suggest that NF-κB can activate cyclin D1 promoter activity primarily by the proximal binding site interacting with the classical p50/p65 complex (12,13,29). We aimed to ascertain whether HBZ contributes to the regulation of this signaling pathway. Accordingly, we first evaluated the effect of HBZ on the NF-κB signaling pathway using a luciferase assay. We transfected 293T and 293T-HBZ cells with pGL3-CD4/-47 luciferase reporter plasmids with or without p65-expressing vectors. HBZ expression suppressed p65-mediated pGL3-CD4/-47 (NF-κB) activation (Fig. 4A). Next, we sought to identify the region of HBZ responsible for repressing NF-κB activation. To this end, we transfected pGL3-CD4/-47 luciferase reporter plasmids with p65 vectors into HBZ-deletion mutants that were stably expressed in 293T cells and then performed a luciferase assy. Wild-type HBZ markedly downregulated p65-mediated pGL3-CD4/-47 (NF-κB) activation (Fig. 4B). Compared with other mutants, only the HBZ-AD+bZIP mutant exhibited significant suppressive activity.

HBZ represses NF-κB activity by binding to p65

The data described above suggest that HBZ influences cyclin D1 promoter activity via the NF-κB p65 signaling pathway. To further investigate whether HBZ could physically interact with NF-κB p65, and to identify whether the HBZ protein was necessary for binding with the NF-κB p65 protein, we performed a co-immunoprecipitation assay. We found that three mutants (HBZ-ΔAD, HBZ-ΔbZIP and HBZ-AD+bZIP) could bind to p65 (Fig. 5A), demonstrating that at least two of the three domains in HBZ were required for binding between HBZ and p65. However, only HBZ-AD+bZIP suppressed NF-κB activation in the luciferase assay (Fig. 4B), indicating the significance of the AD and bZIP domains for p65 binding. In normal cells, p65 and p50 form heterodimers and bind to the κB site in the cyclin D1 promoter. We next studied the binding affinity of HBZ and p50 by immunoprecipitation. As shown in Fig. 5B, HBZ did not bind to p50, nor did it affect binding between p65 and p50.

Taken together, these observations demonstrate that the HBZ-AD and bZIP domains directly physically associate with NF-κB p65, and this interaction acts to inhibit cyclin D1 expression.

Discussion

Accumulating evidence indicates that the cell cycle is regulated by oncogenes and tumor-suppressor genes, and that cell cycle alterations can occur in response to various carcinogens (30,31), implicating cell cycle regulation in carcinogenesis (32). Normal eukaryotic cells have a well-defined cell cycle that consists of four distinct stages: G1, S, G2 and M. G1 and G2 phases represents gaps between the M-S phase and S-M transitions, respectively. These gaps allow for the repair of DNA damage and replication errors. Moreover, G1 represents a period when many pathways can be engaged to influence cell fate (33). Extracellular changes, such as hypoxia, stress and diverse metabolic responses, are integrated and interpreted during this period. Aberrant control of G1, caused by the activation of many oncogenes or the inactivation of tumor-suppressor genes, plays a critical role in tumorigenesis (33). Cyclin D1 is a key regulator of G1-S transition (34). As a sensor that is activated in response to extracellular changes, cyclin D1 can be induced by growth factors and stress in response to the activation of various signaling pathways, including NF-κB (3537).

The transcription factor NF-κB belongs to the Rel family. Mature dimeric NF-κB proteins can translocate to the nucleus and activate genes involved in apoptosis, cell proliferation and angiogenesis. It can be activated in response to many viral infections, and is thought to be important in the host protective response to viral pathogens. Thus, many viruses have evolved distinct strategies to regulate the activation of NF-κB. Furthermore, activation of the NF-κB signaling pathway has also been reported in various types of cancer cells and is be thought to play an important role in the development and progression of tumors (38,39).

HTLV-1 was first reported in 1980 (18). After transmission, it increases the viral copy number by driving the clonal proliferation of infected cells, which results in the onset of ATL (40,41). In this strategy, Tax is known to play a critical role in increasing the number of HTLV-1-infected cells by promoting proliferation and inhibiting apoptosis (42,43). Tax can activate NF-κB by both classical and alternative pathways via interactions with IKKγ and p100 (44,45). However, Tax can often be inactivated by genetic and epigenetic modifications in ATL (40,46). HBZ, another viral gene which is encoded by the minus strand of the HTLV-1 genome, is expressed in all HTLV-1-infected and ATL cells, and promotes the proliferation of these cells (21). Similar to other viral proteins of oncogenic viruses, recent studies have demonstrated multiple functions of HBZ. For example, HBZ can deregulate multiple cellular signaling pathways, including the classical NF-κB, AP-1, transforming growth factor-β (TGF-β), and the Wnt pathways, which possibly contribute to viral persistence and the clonal expansion of infected cells (22,4749). These findings indicate that HBZ is a critical viral gene involved in HTLV-1-mediated oncogenesis.

The present study demonstrated a function of the retroviral protein HBZ and provides some insights into the general mechanism of the proliferation of viral-infected cells. Our data indicate that HBZ can inhibit the expression of cyclin D1 via repression of cyclin D1 promoter activity. Using cyclin D1 promoter mutant reporter constructs, the luciferase assay showed that HBZ downregulated cyclin D1 promoter activity through interactions with the CREB and NF-κB binding sites (Fig. 3), suggesting that HBZ likely interacts with these transcription factors. Our previous study showed that HBZ could directly bind to CREB via a bZIP domain and inhibit CREB transcriptional activity at the CRE site, thereby downregulating levels of cyclin D1 transcription (23). Therefore, in the present study, we further explored the interaction between HBZ and NF-κB. We found that HBZ suppressed activation of the cyclin D1 promoter, which contained an NF-κB binding site, in a manner induced by p65. This suppression was found to occur through the HBZ-AD and bZIP domains (Fig. 4). This finding suggests that HBZ exerts effects on the classical NF-κB signaling pathway, although not all of the three HBZ domains participate in this process, in accord with a previous study (22). Finally, our co-immunoprecipitation findings confirmed that HBZ could bind to p65 via the HBZ-AD and/or bZIP domains in vivo (Fig. 5A). However, HBZ was unable to bind to a single p50 molecule, and binding between HBZ and p65 did not disturb the formation of p65/p50 heterodimers (Fig. 5B).

As HBZ has been found to be an important viral protein, it remains unclear how HTLV-1 induces transformation of T cells into ATL. One viral protein, Tax, induces the expression of many genes and causes the aberrant expression of many cellular genes involved in the growth and survival of T cells (50). However, another viral protein, HBZ, also interacts with many transcriptional factors and exerts an effect that is opposite to Tax on the regulation of cellular signaling pathways. As shown in the present study, HBZ functions as a suppressor of cyclin D1. In accord with previous studies, we identified a mechanism involved in the cell cycle regulation mediated by HBZ. Although Tax has been found to activate cyclin D1 expression and the NF-κB pathway (45,51,52). we believe that HTLV-1 may take advantage of this opposite function of Tax and HBZ in the regulation of signaling pathways to allow for better survival and regulation of the proliferation of HTLV-1-infected cells.

Acknowledgments

The present study was supported by the Foundation for University Key Teacher by the Education Department Henan Province (no. 2015GGJS-224) and the Research Fund for the Key Program of Higher Education of Henan Province (no. 16A310026).

References

1 

Verma IM, Stevenson JK, Schwarz EM, Van Antwerp D and Miyamoto S: Rel/NF-kappa B/I kappa B family: Intimate tales of association and dissociation. Genes Dev. 9:2723–2735. 1995. View Article : Google Scholar : PubMed/NCBI

2 

Baeuerle PA and Baltimore D: NF-kappa B: Ten years after. Cell. 87:13–20. 1996. View Article : Google Scholar : PubMed/NCBI

3 

Baldwin AS Jr, Azizkhan JC, Jensen DE, Beg AA and Coodly LR: Induction of NF-kappa B DNA-binding activity during the G0-to-G1 transition in mouse fibroblasts. Mol Cell Biol. 11:4943–4951. 1991. View Article : Google Scholar : PubMed/NCBI

4 

Chen FE and Ghosh G: Regulation of DNA binding by Rel/NF-kappaB transcription factors: Structural views. Oncogene. 18:6845–6852. 1999. View Article : Google Scholar : PubMed/NCBI

5 

Schmitz ML and Baeuerle PA: The p65 subunit is responsible for the strong transcription activating potential of NF-kappa B. EMBO J. 10:3805–3817. 1991.PubMed/NCBI

6 

Karin M, Cao Y, Greten FR and Li ZW: NF-kappaB in cancer: From innocent bystander to major culprit. Nat Rev Cancer. 2:301–310. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Li Q and Verma IM: NF-kappaB regulation in the immune system. Nat Rev Immunol. 2:725–734. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Hayden MS and Ghosh S: Signaling to NF-kappaB. Genes Dev. 18:2195–2224. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Duyao MP, Buckler AJ and Sonenshein GE: Interaction of an NF-kappa B-like factor with a site upstream of the c-myc promoter. Proc Natl Acad Sci USA. 87:4727–4731. 1990. View Article : Google Scholar : PubMed/NCBI

10 

Kessler DJ, Spicer DB, La Rosa FA and Sonenshein GE: A novel NF-kappa B element within exon 1 of the murine c-myc gene. Oncogene. 7:2447–2453. 1992.PubMed/NCBI

11 

Feng B, Cheng S, Hsia CY, King LB, Monroe JG and Liou HC: NF-kappaB inducible genes BCL-X and cyclin E promote immature B-cell proliferation and survival. Cell Immunol. 232:9–20. 2004. View Article : Google Scholar

12 

Guttridge DC, Albanese C, Reuther JY, Pestell RG and Baldwin AS Jr: NF-kappaB controls cell growth and differentiation through transcriptional regulation of cyclin D1. Mol Cell Biol. 19:5785–5799. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Hinz M, Krappmann D, Eichten A, Heder A, Scheidereit C and Strauss M: NF-kappaB function in growth control: Regulation of cyclin D1 expression and G0/G1-to-S-phase transition. Mol Cell Biol. 19:2690–2698. 1999. View Article : Google Scholar : PubMed/NCBI

14 

Zhou A, Scoggin S, Gaynor RB and Williams NS: Identification of NF-kappa B-regulated genes induced by TNFalpha utilizing expression profiling and RNA interference. Oncogene. 22:2054–2064. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Sherr CJ: Mammalian G1 cyclins. Cell. 73:1059–1065. 1993. View Article : Google Scholar : PubMed/NCBI

16 

Beijersbergen RL and Bernards R: Cell cycle regulation by the retinoblastoma family of growth inhibitory proteins. Biochim Biophys Acta. 1287:103–120. 1996.PubMed/NCBI

17 

Joyce D, Bouzahzah B, Fu M, Albanese C, D'Amico M, Steer J, Klein JU, Lee RJ, Segall JE, Westwick JK, et al: Integration of Rac-dependent regulation of cyclin D1 transcription through a nuclear factor-kappaB-dependent pathway. J Biol Chem. 274:25245–25249. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Poiesz BJ, Ruscetti FW, Gazdar AF, Bunn PA, Minna JD and Gallo RC: Detection and isolation of type C retrovirus particles from fresh and cultured lymphocytes of a patient with cutaneous T-cell lymphoma. Proc Natl Acad Sci USA. 77:7415–7419. 1980. View Article : Google Scholar : PubMed/NCBI

19 

Yoshida M, Miyoshi I and Hinuma Y: Isolation and characterization of retrovirus from cell lines of human adult T-cell leukemia and its implication in the disease. Proc Natl Acad Sci USA. 79:2031–2035. 1982. View Article : Google Scholar : PubMed/NCBI

20 

Kannian P and Green PL: Human T lymphotropic virus type 1 (HTLV-1): Molecular biology and oncogenesis. Viruses. 2:2037–2077. 2010. View Article : Google Scholar

21 

Satou Y, Yasunaga J, Yoshida M and Matsuoka M: HTLV-I basic leucine zipper factor gene mRNA supports proliferation of adult T cell leukemia cells. Proc Natl Acad Sci USA. 103:720–725. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Zhao T, Yasunaga J, Satou Y, Nakao M, Takahashi M, Fujii M and Matsuoka M: Human T-cell leukemia virus type 1 bZIP factor selectively suppresses the classical pathway of NF-kappaB. Blood. 113:2755–2764. 2009. View Article : Google Scholar

23 

Ma Y, Zheng S, Wang Y, Zang W, Li M, Wang N, Li P, Jin J, Dong Z and Zhao G: The HTLV-1 HBZ protein inhibits cyclin D1 expression through interacting with the cellular transcription factor CREB. Mol Biol Rep. 40:5967–5975. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Schmittgen TD and Livak KJ: Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 3:1101–1108. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Gaudray G, Gachon F, Basbous J, Biard-Piechaczyk M, Devaux C and Mesnard JM: The complementary strand of the human T-cell leukemia virus type 1 RNA genome encodes a bZIP transcription factor that downregulates viral transcription. J Virol. 76:12813–12822. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Hivin P, Frédéric M, Arpin-André C, Basbous J, Gay B, Thébault S and Mesnard JM: Nuclear localization of HTLV-I bZIP factor (HBZ) is mediated by three distinct motifs. J Cell Sci. 118:1355–1362. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Zhao T and Matsuoka M: HBZ and its roles in HTLV-1 oncogenesis. Front Microbiol. 3:2472012. View Article : Google Scholar : PubMed/NCBI

28 

Guo ZY, Hao XH, Tan FF, Pei X, Shang LM, Jiang XL and Yang F: The elements of human cyclin D1 promoter and regulation involved. Clin Epigenetics. 2:63–76. 2011. View Article : Google Scholar

29 

Dahlman JM, Wang J, Bakkar N and Guttridge DC: The RelA/p65 subunit of NF-kappaB specifically regulates cyclin D1 protein stability: Implications for cell cycle withdrawal and skeletal myogenesis. J Cell Biochem. 106:42–51. 2009. View Article : Google Scholar

30 

Sgambato A, Han EK, Zhang YJ, Moon RC, Santella RM and Weinstein IB: Deregulated expression of cyclin D1 and other cell cycle-related genes in carcinogen-induced rat mammary tumors. Carcinogenesis. 16:2193–2198. 1995. View Article : Google Scholar : PubMed/NCBI

31 

Lee CC, Yamamoto S, Wanibuchi H, Wada S, Sugimura K, Kishimoto T and Fukushima S: Cyclin D1 overexpression in rat two-stage bladder carcinogenesis and its relationship with oncogenes, tumor suppressor genes, and cell proliferation. Cancer Res. 57:4765–4776. 1997.PubMed/NCBI

32 

Sherr CJ: Cancer cell cycles. Science. 274:1672–1677. 1996. View Article : Google Scholar : PubMed/NCBI

33 

Massagué J: G1 cell-cycle control and cancer. Nature. 432:298–306. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Semczuk A and Jakowicki JA: Alterations of pRb1-cyclin D1-cdk4/6-p16(INK4A) pathway in endometrial carcinogenesis. Cancer Lett. 203:1–12. 2004. View Article : Google Scholar

35 

Watanabe G, Albanese C, Lee RJ, Reutens A, Vairo G, Henglein B and Pestell RG: Inhibition of cyclin D1 kinase activity is associated with E2F-mediated inhibition of cyclin D1 promoter activity through E2F and Sp1. Mol Cell Biol. 18:3212–3222. 1998. View Article : Google Scholar : PubMed/NCBI

36 

Lee RJ, Albanese C, Stenger RJ, Watanabe G, Inghirami G, Haines GK III, Webster M, Muller WJ, Brugge JS, Davis RJ, et al: pp60(v-src) induction of cyclin D1 requires collaborative interactions between the extracellular signal-regulated kinase, p38, and Jun kinase pathways. A role for cAMP response element-binding protein and activating transcription factor-2 in pp60(v-src) signaling in breast cancer cells. J Biol Chem. 274:7341–7350. 1999. View Article : Google Scholar : PubMed/NCBI

37 

Ozes ON, Mayo LD, Gustin JA, Pfeffer SR, Pfeffer LM and Donner DB: NF-kappaB activation by tumour necrosis factor requires the Akt serine-threonine kinase. Nature. 401:82–85. 1999. View Article : Google Scholar : PubMed/NCBI

38 

Pikarsky E, Porat RM, Stein I, Abramovitch R, Amit S, Kasem S, Gutkovich-Pyest E, Urieli-Shoval S, Galun E and Ben-Neriah Y: NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature. 431:461–466. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Karin M: Nuclear factor-kappaB in cancer development and progression. Nature. 441:431–436. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Matsuoka M and Jeang KT: Human T-cell leukaemia virus type 1 (HTLV-1) infectivity and cellular transformation. Nat Rev Cancer. 7:270–280. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Matsuoka M: Human T-cell leukemia virus type I (HTLV-I) infection and the onset of adult T-cell leukemia (ATL). Retrovirology. 2:272005. View Article : Google Scholar : PubMed/NCBI

42 

Mulloy JC, Kislyakova T, Cereseto A, Casareto L, LoMonico A, Fullen J, Lorenzi MV, Cara A, Nicot C, Giam C, et al: Human T-cell lymphotropic/leukemia virus type 1 Tax abrogates p53-induced cell cycle arrest and apoptosis through its CREB/ATF functional domain. J Virol. 72:8852–8860. 1998.PubMed/NCBI

43 

Yoshida M: Multiple viral strategies of HTLV-1 for dysregulation of cell growth control. Annu Rev Immunol. 19:475–496. 2001. View Article : Google Scholar : PubMed/NCBI

44 

Chu ZL, Shin YA, Yang JM, DiDonato JA and Ballard DW: IKKgamma mediates the interaction of cellular IkappaB kinases with the tax transforming protein of human T-cell leukemia virus type 1. J Biol Chem. 274:15297–15300. 1999. View Article : Google Scholar : PubMed/NCBI

45 

Xiao G, Cvijic ME, Fong A, Harhaj EW, Uhlik MT, Waterfield M and Sun SC: Retroviral oncoprotein Tax induces processing of NF-kappaB2/p100 in T-cells: Evidence for the involvement of IKKalpha. EMBO J. 20:6805–6815. 2001. View Article : Google Scholar : PubMed/NCBI

46 

Kannagi M, Harada S, Maruyama I, Inoko H, Igarashi H, Kuwashima G, Sato S, Morita M, Kidokoro M, Sugimoto M, et al: Predominant recognition of human T-cell leukemia virus type I (HTLV-I) pX gene products by human CD8+ cytotoxic T cells directed against HTLV-I-infected cells. Int Immunol. 3:761–767. 1991. View Article : Google Scholar : PubMed/NCBI

47 

Matsumoto J, Ohshima T, Isono O and Shimotohno K: HTLV-1 HBZ suppresses AP-1 activity by impairing both the DNA-binding ability and the stability of c-Jun protein. Oncogene. 24:1001–1010. 2005. View Article : Google Scholar

48 

Zhao T, Satou Y, Sugata K, Miyazato P, Green PL, Imamura T and Matsuoka M: HTLV-1 bZIP factor enhances TGF-β signaling through p300 coactivator. Blood. 118:1865–1876. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Ma G, Yasunaga J, Fan J, Yanagawa S and Matsuoka M: HTLV-1 bZIP factor dysregulates the Wnt pathways to support proliferation and migration of adult T-cell leukemia cells. Oncogene. 32:4222–4230. 2013. View Article : Google Scholar

50 

Ressler S, Connor LM and Marriott SJ: Cellular transformation by human T-cell leukemia virus type I. FEMS Microbiol Lett. 140:99–109. 1996. View Article : Google Scholar : PubMed/NCBI

51 

Kim YM, Geiger TR, Egan DI, Sharma N and Nyborg JK: The HTLV-1 tax protein cooperates with phosphorylated CREB, TORC2 and p300 to activate CRE-dependent cyclin D1 transcription. Oncogene. 29:2142–2152. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Harhaj NS, Sun SC and Harhaj EW: Activation of NF-kappa B by the human T cell leukemia virus type I Tax oncoprotein is associated with ubiquitin-dependent relocalization of I kappa B kinase. J Biol Chem. 282:4185–4192. 2007. View Article : Google Scholar

Related Articles

Journal Cover

March-2017
Volume 39 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ma Y, Zhang B, Wang D, Qian L, Song X, Wang X, Yang C and Zhao G: HTLV-1 basic leucine zipper factor downregulates cyclin D1 expression via interactions with NF-κB. Int J Mol Med 39: 764-770, 2017
APA
Ma, Y., Zhang, B., Wang, D., Qian, L., Song, X., Wang, X. ... Zhao, G. (2017). HTLV-1 basic leucine zipper factor downregulates cyclin D1 expression via interactions with NF-κB. International Journal of Molecular Medicine, 39, 764-770. https://doi.org/10.3892/ijmm.2017.2868
MLA
Ma, Y., Zhang, B., Wang, D., Qian, L., Song, X., Wang, X., Yang, C., Zhao, G."HTLV-1 basic leucine zipper factor downregulates cyclin D1 expression via interactions with NF-κB". International Journal of Molecular Medicine 39.3 (2017): 764-770.
Chicago
Ma, Y., Zhang, B., Wang, D., Qian, L., Song, X., Wang, X., Yang, C., Zhao, G."HTLV-1 basic leucine zipper factor downregulates cyclin D1 expression via interactions with NF-κB". International Journal of Molecular Medicine 39, no. 3 (2017): 764-770. https://doi.org/10.3892/ijmm.2017.2868